Papanicolaou Test For Ovarian And Endometrial Cancers

Kinde; Isaac ;   et al.

Patent Application Summary

U.S. patent application number 14/439041 was filed with the patent office on 2015-10-15 for papanicolaou test for ovarian and endometrial cancers. This patent application is currently assigned to THE JOHNS HOPKINS UNIVERSITY. The applicant listed for this patent is THE JOHNS HOPKINS UNIVERSITY. Invention is credited to Chetan Bettegowda, Luis Diaz, Isaac Kinde, Kenneth W. Kinzler, Nickolas Papadopoulos, Bert Vogelstein, Yuxuan Wang.

Application Number20150292027 14/439041
Document ID /
Family ID50627940
Filed Date2015-10-15

United States Patent Application 20150292027
Kind Code A1
Kinde; Isaac ;   et al. October 15, 2015

PAPANICOLAOU TEST FOR OVARIAN AND ENDOMETRIAL CANCERS

Abstract

The recently developed liquid-based Papanicolaou (Pap) smear allows not only cytologic evaluation but also collection of DNA for detection of HPV, the causative agent of cervical cancer. We tested these samples to detect somatic mutations present in rare tumor cells that might accumulate in the cervix once shed from endometrial and ovarian cancers. A panel of commonly mutated genes in endometrial and ovarian cancers was assembled and used to identify mutations in all 46 endometrial or cervical cancer tissue samples. We were able also able to identify the same mutations in the DNA from liquid Pap smears in 100% of endometrial cancers (24 of 24) and in 41% of ovarian cancers (9 of 22). We developed a sequence-based method to query mutations in 12 genes in a single liquid Pap smear without prior knowledge of the tumor's genotype.


Inventors: Kinde; Isaac; (Beaumont, CA) ; Kinzler; Kenneth W.; (Baltimore, MD) ; Vogelstein; Bert; (Baltimore, MD) ; Papadopoulos; Nickolas; (Towson, MD) ; Diaz; Luis; (Ellicott City, MD) ; Bettegowda; Chetan; (Perry Hall, MD) ; Wang; Yuxuan; (Baltimore, MD)
Applicant:
Name City State Country Type

THE JOHNS HOPKINS UNIVERSITY

Baltimore

MD

US
Assignee: THE JOHNS HOPKINS UNIVERSITY
Baltimore
MD

Family ID: 50627940
Appl. No.: 14/439041
Filed: October 17, 2013
PCT Filed: October 17, 2013
PCT NO: PCT/US2013/065342
371 Date: April 28, 2015

Related U.S. Patent Documents

Application Number Filing Date Patent Number
61719942 Oct 29, 2012

Current U.S. Class: 506/2 ; 435/29; 435/6.11; 435/7.1; 435/7.23; 435/7.4; 435/7.92; 506/16; 506/9
Current CPC Class: G01N 33/57449 20130101; C12Q 2600/156 20130101; C12Q 2600/154 20130101; C12Q 2600/16 20130101; C12Q 2600/158 20130101; G01N 33/57442 20130101; C12Q 1/6886 20130101
International Class: C12Q 1/68 20060101 C12Q001/68

Goverment Interests



[0001] This invention was made using funds from the National Cancer Institute and the National Institutes of Health. The U.S. government retains certain rights under the terms of NCI contract N01-CN-43309 and NIH grants CA129825 and CA43460.
Claims



1. A method comprising: testing a specimen comprising cells or cell fragments collected from the gynecological tract of a human subject for a genetic or epigenetic change in one or more genes, mRNAs, or proteins mutated in endometrial, fallopian tubal, or ovarian cancer.

2. The method of claim 1 wherein the change is a substitution mutation.

3. The method of claim 1 wherein the change is a rearrangement.

4. The method of claim 1 wherein the change is a deletion.

5. The method of claim 1 wherein the change is a loss or gain of methylation.

6. The method of claim 1 wherein the change is determined with respect to the bulk of the genes, mRNAs, or proteins present in the specimen.

7. A method comprising: testing a specimen comprising cells or cell fragments collected from the gynecological tract of a human subject for one or more mutations in a gene, mRNA, or protein selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

8. The method of claim 7 wherein the step of testing is performed on at least 3 of said genes, mRNAs, or proteins.

9. The method of claim 7 wherein the step of testing is performed on at least 5 of said genes, mRNAs, or proteins.

10. The method of claim 7 wherein the step of testing is performed on at least 7 of said genes, mRNAs, or proteins.

11. The method of claim 7 wherein the step of testing is performed on at least 9 of said genes, mRNAs, or proteins.

12. The method of claim 7 wherein the step of testing is performed on at least 11 of said genes, mRNAs, or proteins.

13. The method of claim 7 wherein the step of testing is performed on at least 12 of said genes, mRNAs, or proteins.

14. The method of claim 7 wherein the step of testing is performed in a multiplex assay.

15. The method of claim 7 wherein the step of testing is repeated over time.

16. The method of claim 7 wherein the liquid Pap specimen is collected after surgical debulking of an ovarian tumor.

17. A kit for testing a panel of genes in Pap specimens for ovarian or endometrial cancers, the kit comprising a at least 10 probes or at least 10 primer pairs, wherein each probe or primer comprises at least 15 nt of complementary sequence to one of the panel of genes, wherein the panel is cumulatively complementary to at least 10 different genes, wherein the panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

18. The kit of claim 17 which comprises probes and wherein the probes are attached to a solid support.

19. The kit of claim 17 which comprises primer pairs, wherein the primer pairs prime synthesis of a nucleic acid of between 240 and 300 bp.

20. The kit of claim 17 which comprises primer pairs, wherein the primer pairs prime synthesis of a nucleic acid of between 200 and 325 bp.

21. The kit of claim 17 which comprises primer pairs, wherein the primer pairs prime synthesis of a nucleic acid of between 60 and 1000 bp.

22. The kit of claim 21 wherein at least one primer from each primer pair is attached to a solid support.

23. The kit of claim 17 wherein the probe or primer comprises at least 20 nt of complementary sequence to one of the panel of genes.

24. A solid support comprising at least 10 probes attached thereto, wherein each probe comprises at least 15 nt of complementary sequence to one of a panel of genes, wherein the panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2, wherein the panel is cumulatively complementary to at least 10 different genes.

25. A solid support comprising at least 10 primers attached thereto, wherein each primer comprises at least 15 nt of complementary sequence to one of a panel of genes, wherein the panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2, wherein the panel is cumulatively complementary to at least 10 different genes.

26. The method of claim 1 wherein the specimen is a liquid Pap specimen.

27. The method of claim 7 wherein the specimen is a liquid Pap specimen.

28. The method of claim 1 wherein the specimen is collected from the cervix.

29. The method of claim 7 wherein the specimen is collected from the cervix.

30. The method of claim 7 wherein the step of testing is performed by increasing the sensitivity of massively parallel sequencing instruments with an error reduction technique that allows for the detection of rare mutant alleles in a range of 1 mutant template among 5,000 to 1,000,000 wild-type templates.

31. The method of claim 7 wherein the step of testing is performed by increasing the sensitivity of massively parallel sequencing instruments with an error reduction technique that includes: a. assignment of a unique identifier (UID) to each template molecule; b. amplification of each uniquely tagged template molecule to create UID-families; and c. redundant sequencing of the amplification products.
Description



TECHNICAL FIELD OF THE INVENTION

[0002] This invention is related to the area of cancer screening. In particular, it relates to ovarian and endometrial cancers.

BACKGROUND OF THE INVENTION

[0003] Since the introduction of the Papanicolaou test, the incidence and mortality of cervical cancer in screened populations has been reduced by more than 75% (1, 2). In contrast, deaths from ovarian and endometrial cancers have not substantially decreased during that same time period. As a result, more than 69,000 women in the U.S. will be diagnosed with ovarian and endometrial cancer in 2012. Although endometrial cancer is more common than ovarian cancer, the latter is more lethal. In the U.S., approximately 15,000 and 8,000 women are expected to die each year from ovarian and endometrial cancers, respectively (Table 1). World-wide, over 200,000 deaths from these tumors are expected this year alone (3, 4).

[0004] In an effort to replicate the success of cervical cancer screening, several approaches for the early detection of endometrial and ovarian cancers have been devised. For endometrial cancers, efforts have focused on cytology and transvaginal ultrasound (TVS). Cytology can indeed indicate a neoplasm within the uterus in some cases, albeit with low specificity (5). TVS is a noninvasive technique to measure the thickness of the endometrium based on the fact that endometria harboring a cancer are thicker than normal endometria (6). As with cytology, screening measurement of the endometrial thickness using TVS lacks sufficient specificity because benign lesions, such as polyps, can also result in a thickened endometrium. Accordingly, neither cytology nor TVS fulfills the requirements for a screening test (5, 7).

[0005] Even greater efforts have been made to develop a screening test for ovarian cancer, using serum CA-125 levels and TVS. CA-125 is a high molecular weight transmembrane glycoprotein expressed by coelomic- and Mullerian-derived epithelia that is elevated in a subset of ovarian cancer patients with early stage disease (8). The specificity of CA-125 is limited by the fact that it is also elevated in a variety of benign conditions, such as pelvic inflammatory disease, endometriosis and ovarian cysts (9). TVS can visualize the ovary but can only detect large tumors and cannot definitively distinguish benign from malignant tumors. Several clinical screening trials using serum CA-125 and TVS have been conducted but none has shown a survival benefit. In fact, some have shown an increase in morbidity compared to controls because false positive tests elicit further evaluation by laparoscopy or exploratory laparotomy (10-12).

[0006] Accordingly, the U.S. Preventative Services Task Force, the American Cancer Society, the American Congress of Obstetricians and Gynecologists, as well as the National Comprehensive Cancer Network, do not recommend routine screening for endometrial or ovarian cancers in the general population. In fact, these organizations warn that "the potential harms outweigh the potential benefits" (13-16). An exception to this recommendation has been made for patients with a hereditary predisposition to ovarian cancer, such as those with germline mutations in a BRCA gene or those with Lynch syndrome. It is recommended that BRCA mutation carriers be screened every 6 months with TVS and serum CA-125, starting at a relatively early age. Screening guidelines for women with Lynch syndrome include annual endometrial sampling and TVS beginning between age 30 and 35 (15, 17).

[0007] The mortality associated with undetected gynecologic malignancies has made the development of an effective screening tool a high priority. An important observation that inspired the current study is that asymptomatic women occasionally present with abnormal glandular cells (AGCs) detected in a cytology specimen as part of their routine cervical cancer screening procedure. Although AGCs are associated with premalignant or malignant disease in some cases (18-22), it is often difficult to distinguish the AGCs arising from endocervical, endometrial or ovarian cancer from one another or from more benign conditions. There is a continuing need in the art to detect these cancers at an earlier stage than done currently.

SUMMARY OF THE INVENTION

[0008] According to one aspect of the invention a method is provided for detecting or monitoring endometrial or ovarian cancer. A liquid Pap smear of a patient is tested for a genetic or epigenetic change in one or more genes, mRNAs, or proteins mutated in endometrial or ovarian cancer. Detection of the change indicates the presence of such a cancer in the patient.

[0009] According to another aspect of the invention a method is provided for screening for endometrial and ovarian cancers. A liquid Pap smear is tested for one or more mutations in a gene, mRNA, or protein selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2. Detection of the mutation indicates the presence of such a cancer in the patient.

[0010] Another aspect of the invention is a kit for testing a panel of genes in Pap smear samples for ovarian or endometrial cancers. The kit comprises at least 10 probes or at least 10 primer pairs. Each probe or primer comprises at least 15 nt of complementary sequence to one of the panel of genes. At least 10 different genes are interrogated. The panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

[0011] Still another aspect of the invention is a solid support comprising at least 10 attached probes. Each probe comprises at least 15 nt of complementary sequence to one of a panel of genes, wherein the panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

[0012] Another aspect of the invention is a solid support comprising at least 10 primers attached thereto. Each primer comprises at least 15 nt of complementary sequence to one of a panel of genes. The panel is selected from the group consisting of CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

[0013] These and other embodiments which will be apparent to those of skill in the art upon reading the specification provide the art with methods for assessing ovarian and endometrial cancers in a screening environment using samples that are already routinely collected.

BRIEF DESCRIPTION OF THE DRAWINGS

[0014] FIG. 1. Schematic demonstrating the principle steps of the procedure described in this study. Tumors cells shed from ovarian or endometrial cancers are carried into the endocervical canal. These cells can be captured by the brush used for performing a routine Pap smear. The brush contents are transferred into a liquid fixative, from which DNA is isolated. Using next-generation sequencing, this DNA is queried for mutations that indicate the presence of a malignancy in the female reproductive tract.

[0015] FIG. 2. Diagram of the assay used to simultaneously detect mutations in 12 different genes. A modification of the Safe-SeqS (Safe-Sequencing System) protocol, for simultaneous interrogation of multiple mutations in a single sample, is depicted. In the standard Safe-SeqS procedure, only one amplicon is assessed, while the new system is used to assess multiple amplicons from multiple genes at once.

[0016] FIG. 3. Mutant allele fractions in Pap smear fluids. The fraction of mutant alleles from each of 46 pap smear fluids is depicted. The stage of each tumor is listed on the Y-axis. The X-axis demonstrates the % mutant allele fraction as determined by Safe-SeqS.

[0017] FIG. 4. Heat map depicting the results of multiplex testing of 12 genes in Pap smear fluids. Each block on the y-axis represents a 30-bp block of sequence from the indicated gene. The 28 samples assessed (14 from women with cancer, 14 from normal women without cancer) are indicated on the x-axis. Mutations are indicated as colored blocks, with white indicating no mutation, yellow indicating a mutant fraction of 0.1% to 1%, orange indicate a mutant fraction of 1% to 10%, and red indicating a mutant fraction of >10%.

[0018] FIG. 5. Table 1. Epidemiology of Ovarian and Endometrial Tumors. The estimated numbers of new cases and deaths in the U.S. from the major subtypes of ovarian and endometrial cancers are listed.

[0019] FIG. 6. Table 2. Genetic Characteristics of Ovarian and Endometrial Cancers. The frequencies of the commonly mutated genes in ovarian and endometrial cancers are listed.

[0020] FIG. 7. Table S1. Endometrial Cancers (Endometrioid Subtype) Studied by Whole-exome Sequencing. The summary characteristics of the 22 cancers used for exome sequencing are listed.

[0021] FIG. 8. Table S3. Mutations Assessed in Pap Smears. Clinical characteristics of the 46 tumor samples are listed, along with the mutation identified in each case and the fraction of mutant alleles identified in the Pap smears.

[0022] FIG. 9. Table S4. Primers Used to Assess Individual Mutations in Pap Smears. The sequences of the forward and reverse primers used to test each mutation via Safe-SeqS are listed in pairs (SEQ ID NO: 4-99, respectively).

[0023] FIG. 10. Table S5. Primers Used to Simultaneously Assess 12 Genes in Pap Smears. The sequences of the forward and reverse primers for each tested region are listed in pairs (SEQ ID NO: 100-191, respectively).

[0024] FIG. 11. Table S6. Mutations Identified in Pap Smears through Simultaneous Assessment of 12 Genes. The fraction of mutant alleles identified in the Pap smears using this approach is listed, along with the precise mutations identified.

DETAILED DESCRIPTION OF THE INVENTION

[0025] The inventors have developed a test for detecting different cancers using samples that are already routinely collected for diagnosing uterine cancer and HPV (human papilloma virus) infection. Using a panel of genes, a high level of detection of both endometrial and ovarian cancers was achieved.

[0026] Certain genes have been identified as mutated in a high proportion of endometrial and ovarian cancers. These include CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2. The test can be performed on at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 of these genes. In addition, other genes can be added or substituted into the panel to achieve a higher rate of detection.

[0027] Testing for a mutation may be done by analysis of nucleic acids, such as DNA or mRNA or cDNA. The nucleic acid analytes are isolated from cells or cell fragments found in the liquid PAP smear sample. Suitable tests may include any hybridization or sequencing based assay. Analysis may also be performed on protein encoded by the genes in the panel. Any suitable test may be used including but not limited to mass spectrometry. Other suitable assays may include immunological assays, such as, immunoblotting, immunocytochemistry, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoradiometric assays (IRMA) and immunoenzymatic assays (IEMA), including sandwich assays using monoclonal or polyclonal antibodies.

[0028] Genetic changes which can be detected are typically mutations such as deletions, insertions, duplications, substitutions (missense or nonsense mutations), rearrangements, etc. Such mutations can be detected inter alia by comparing to a wild type in another (non-tumor) tissue or fluid of an individual or by comparing to reference sequences, for example in databases. Mutations that are found in all tissues of an individual are germline mutations, whereas those that occur only in a single tissue are somatic mutations. Epigenetic changes can also be detected. These may be loss or gain of methylation at specific locations in specific genes, as well as histone modifications, including acetylation, ubiquitylation, phosphorylation and sumoylation.

[0029] Tests may be done in a multiplex format, in which a single reaction pot is used to detect multiple analytes. Examples of such tests include amplifications using multiple primer sets, amplifications using universal primers, array based hybridization or amplification, emulsion based amplification.

[0030] While probes and primers may be designed to interrogate particular mutations or particular portions of a gene, mRNA, or cDNA, these may not be separate entities. For example, probes and primers may be linked together to form a concatamer, or they may be linked to one or more solid supports, such as a bead or an array.

[0031] Kits for use in the disclosed methods may include a carrier for the various components. The carrier can be a container or support, in the form of, e.g., bag, box, tube, rack, and is optionally compartmentalized. The kit also includes various components useful in detecting mutations, using the above-discussed detection techniques. For example, the detection kit may include one or more oligonucleotides useful as primers for amplifying all or a portion of the target nucleic acids. The detection kit may also include one or more oligonucleotide probes for hybridization to the target nucleic acids. Optionally the oligonucleotides are affixed to a solid support, e.g., incorporated in a microarray included in the kit or supplied separately.

[0032] Solid supports may contain one single primer or probe or antibody for detecting a single gene, protein, mRNA, or portion of a gene. A solid support may contain multiple primers, probes, or antibodies. They may be provided as a group which will interrogate mutations at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 of the genes of the desired panel. The panel may be selected from or comprise CTNNB1, EGFR, PI3KCA, PTEN, TP53, BRAF, KRAS, AKT1, NRAS, PPP2R1A, APC, FBXW7, ARID1A, CDKN2A, MLL2, RFF43, and FGFR2.

[0033] Primer pairs may be used to synthesize amplicons of various sizes. Amplicons may be for example from 50, 60, 75, 100, 125, 150, 200, 140, 180 bp in length. Amplicons may run up to 200, 250, 300, 400, 500, 750, 1000 bp in length, as examples. The size of the amplicon may be limited by the size and/or quality of the template retrieved from the liquid PAP smear. Probes and primers for use in the invention may contain a wild-type sequence or may contain a sequence of a particular mutant.

[0034] In one embodiment, the test can be performed using samples that are collected over time. The test results can be compared for quantitative or qualitative changes. Such analysis can be used after a potentially curative therapy, such as surgery.

[0035] Georgios Papanicolaou published his seminal work, entitled "Diagnosis of Uterine Cancer by the Vaginal Smear," in 1943 (31). At that time, he suggested that endocervical sampling could, in theory, be used to detect not only cervical cancers but also other cancers arising in the female reproductive tract, including endometrial carcinomas. The research reported here moves us much closer to that goal. In honor of Papanicolaou's pioneering contribution to the field of early cancer detection, we have named the approach described herein as the "PapGene" test.

[0036] One of the most important developments over the last several years is the recognition that all human cancers are the result of mutations in a limited set of genes and an even more limited set of pathways through which these genes act (32). The whole-exome sequencing data we present, combined with previous genome-wide studies, provide a striking example of the common genetic features of cancer (FIG. 6, Table 2). Through the analysis of particular regions of only 12 genes (FIG. 11, table S5), we could detect at least one driver mutation in the vast majority of nine different gynecologic cancers (FIG. 5, Table 1). Though several of these 12 genes were tumor suppressors, and therefore difficult to therapeutically target, knowledge of their mutational patterns provides unprecedented opportunities for cancer diagnostics.

[0037] The most important finding in this paper is that significant amounts of cells or cell fragments from endometrial and ovarian cancers are present in the cervix and can be detected through molecular genetic approaches. Detection of malignant cells from endometrial and ovarian carcinomas in cervical cytology specimens is relatively uncommon. Microscopic examination cannot always distinguish them from one another, from cervical carcinomas, or from more benign conditions. Our study showed that 100% of endometrial cancers (n=24), even those of low grade, and 41% of ovarian cancers (n=22), shed cells into the cervix that could be detected from specimens collected as part of routine Pap smears. This finding, in conjunction with technical advances allowing the reliable detection of mutations present in only a very small fraction of DNA templates, provided the foundation for the PapGene test.

[0038] This study demonstrates the value of sensitive endocervical DNA testing but there are many issues that need to be addressed before optimal clinical use is achieved. The test, even in its current format, appears to be promising for screening endometrial cancer, as the data in FIG. 3 show that even the lowest stage endometrial cancers could be detected through the analysis of DNA in Pap smear fluid through Safe-SeqS. However, only 41% of ovarian cancers could be detected in Pap smears even when the mutations in their tumors were known. In eight of the nine Pap smears from ovarian cancer patients that contained detectable mutations, the mutant allele fractions were >0.1% and therefore within the range currently detectable by PapGene testing (FIG. 9, table S3). Further improvements in the technology could increase the technical sensitivity of the PapGene test and allow it to detect more ovarian cancers. Other strategies to increase this sensitivity involve physical maneuvers, such as massaging the adnexal region during the pelvic examination or by performing the PapGene test at specified times during the menstrual cycle. Development of an improved method of collection may also be able to improve sensitivity. The current liquid specimen is designed for the detection of cervical cancer and as such utilizes a brush that collects cells from the ectocervix and only minimally penetrates the endocervical canal. A small cannula that can be introduced into the endometrial cavity similar to the pipelle endometrial biopsy instrument could theoretically obtain a more enriched sample of cells coming from the endometrium, fallopian tube and ovary (33).

[0039] The high sensitivity and the quantitative nature of the PapGene test also opens the possibility of utilizing it to monitor the response to hormonal agents (e.g., progestins) used to treat young women with low risk endometrial cancers. Some of these women choose to preserve fertility, undergoing medical therapy rather than hysterectomy (34). The detection of pre-symptomatic ovarian cancers, even if advanced, could also be of benefit. Although not entirely analogous, it has been demonstrated that one of the most important prognostic indicators for ovarian cancer is the amount of residual disease after surgical debulking Initially, debulking was considered optimal if the residual tumor was less than 2 cm. Subsequently, the threshold was reduced to 1 cm and surgeons now attempt to remove any visible tumor. With each improvement in surgical debulking, survival has lengthened (35). A small volume of tumor is likely to be more sensitive to cytotoxic chemotherapy than the large, bulky disease typical of symptomatic high-grade serous carcinoma.

[0040] An essential aspect of the screening approach described here is that it should be relatively inexpensive and easily incorporated into the pelvic examination. Evaluation of HPV DNA is already part of routine Pap smear testing because HPV analysis increases the test's sensitivity (36, 37). The DNA purification component of the PapGene test is identical to that used for HPV, so this component is clearly feasible. The preparation of DNA, multiplex amplification, and the retail cost of the sequencing component of the PapGene test can also be performed at a cost comparable to a routine HPV test in the U.S. today. Note that the increased sensitivity provided by the Safe-SeqS component of the PapGene test (see Example 6) can be implemented on any massively parallel sequencing instrument, not just those manufactured by Illumina. With the reduction in the cost of massively parallel sequencing expected in the future, PapGene testing should become even less expensive.

[0041] There are millions of Pap smear tests performed annually in the U.S. Could PapGene testing be performed on such a large number of specimens? We believe so, because the entire DNA purification and amplification process can be automated, just as it is for HPV testing. Though it may now seem unrealistic to have millions of these sophisticated sequence-based tests performed every year, it would undoubtedly have seemed unrealistic to have widespread, conventional Pap smear testing performed when Papanicolaou published his original paper (31). Even today, when many cervical cytology specimens are screened using automated technologies, a significant percentage require evaluation by a skilled cytopathologist. In contrast, the analysis of PapGene testing is done completely in silico and the read-out of the test is objective and quantitative.

[0042] In sum, PapGene testing has the capacity to increase the utility of conventional cytology screening through the unambiguous detection of endometrial and ovarian carcinomas. In addition to the analysis of much larger numbers of patients with and without various types of endometrial, ovarian, and fallopian tube cancers, the next step in this line of research is to include genes altered in cervical cancer as well as HPV amplicons in the multiplexed Safe-SeqS assay (FIG. 11, table S5). These additions will provide information that could be valuable for the management of patients with the early stages of cervical neoplasia, as HPV positivity alone is not specific for the detection of cervical cancer and its precursor lesions, particularly in young, sexually active women who frequently harbor HPV infections in the absence of neoplasia.

[0043] The above disclosure generally describes the present invention. All references disclosed herein are expressly incorporated by reference. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.

Example 1

[0044] We reasoned that more sophisticated molecular methods might be able to detect the presence of cancer cells in endocervical specimens at higher sensitivities and specificities than possible with conventional methods. In particular, we hypothesized that somatic mutations characteristic of endometrial and ovarian cancers would be found in the DNA purified from routine liquid-based Pap smears (henceforth denoted as "Pap smears"; FIG. 1). Unlike cytologically abnormal cells, such oncogenic DNA mutations are specific, clonal markers of neoplasia that should be absent in non-neoplastic cells. However, we did not know if such DNA would indeed be present in endocervical specimens, and we did not know if they would be present in a sufficient amount to detect them. The experiments described here were carried out to test our hypothesis.

[0045] There were four components to this study: I. Determination of the somatic mutations typically present in endometrial and ovarian cancers; II. Identification of at least one mutation in the tumors of 46 patients with these cancers; III. Determination of whether the mutations identified in these tumors could also be detected in Pap smears from the same patients; and IV. Development of a technology that could directly assess cells from Pap smears for mutations commonly found in endometrial or ovarian cancers.

Example 2

Prevalence of Somatically Mutated Genes in Endometrial and Ovarian Cancers

[0046] There are five major histopathologic subtypes of ovarian cancers. The most prevalent subtype is high grade serous (60% of total), followed by endometrioid (15%), clear cell (10%), and low-grade serous carcinoma (8%) (Table 1). Genome-wide studies have identified the most commonly mutated genes among the most prevalent ovarian cancer subtypes (Table 2) (23-25).

[0047] Such comprehensive studies have not yet been reported for the endometrioid and mucinous subtypes, collectively representing .about.20% of ovarian cancer cases (Table 1). However, commonly mutated genes in the endometrioid and mucinous subtypes have been reported (26). In aggregate, the most commonly mutated gene in epithelial ovarian cancers was TP53, which was mutated in 69% of these cancers (Table 2). Other highly mutated genes included ARID1A, BRAF, CTNNB1, KRAS, PIK3CA, and PPP2R1A (Table 2).

[0048] Among endometrial cancers, the endometrioid subtype is by far the most common, representing 85% of the total (Table 1). Because cancers of this subtype are so frequent and have not been analyzed at a genome-wide level, we evaluated them through whole-exome sequencing. The DNA purified from 22 sporadic endometrioid carcinomas, as well as from matched non-neoplastic tissues, was used to generate 44 libraries suitable for massively parallel sequencing. The clinical aspects of the patients and histopathologic features of the tumors are listed in table S1. Though the examination of 22 cancers cannot provide a comprehensive genome landscape of a tumor type, it is adequate for diagnostic purposes--as these only require the identification of the most frequently mutated genes.

[0049] Among the 44 libraries, the average coverage of each base in the targeted region was 149.1 with 88.4% of targeted bases represented by at least ten reads. Using stringent criteria for the identification of somatic mutations (as described in Materials and Methods), the sequencing data clearly demarcated the tumors into two groups: ten cancers (termed the N Group, for non-highly mutated) harbored <100 somatic mutations per tumor (median 32, range 7 to 50), while 12 cancers (termed the H Group, for highly mutated) harbored >100 somatic mutations per tumor (median 674, range 164 to 4,629) (FIG. 7, table S1).

[0050] The high number of mutations in the Group H tumors was consistent with a deficiency in DNA repair. Eight of the 12 Group H tumors had microsatellite instability (MSI-H, table S1), supporting this conjecture. Moreover, six of the Group H tumors contained somatic mutations in the mismatch repair genes MSH2 or MSH6, while none of the Group N cancers contained mutations in mismatch repair genes. Mismatch repair deficiency is known to be common among endometrial cancers and these tumors occur in 19-71% of women with inherited mutations of mismatch repair genes (i.e., patients with the Hereditary Nonpolyposis Colorectal Cancer) (27).

[0051] 12,795 somatic mutations were identified in the 22 cancers. The most commonly mutated genes included the PIK3 pathway genes PTEN and PIK3CA (28), the APC pathway genes APC and CTNNB1, the fibroblast growth factor receptor FGFR2, the adapter protein FBXW7, and the chromatin-modifying genes ARID1A and MLL2 (Table 2). Genes in these pathways were mutated in both Group N and H tumors. Our results are consistent with prior studies of endometrioid endometrial cancer that had evaluated small numbers of genes, though mutations in FBXW7, MLL2 and APC had not been appreciated to occur as frequently as we found them. It was also interesting that few TP53 mutations (5%) were found in these endometrial cancers (Table 2), a finding also consistent with prior studies.

[0052] Papillary serous carcinomas of the endometrium account for 10-15% of endometrial cancers, and a recent genome-wide sequencing study of this tumor subtype has been published (29). The most common mutations in this subtype are listed in Table 2. The least common subtype of endometrial cancers is clear cell carcinomas, which occur in <5%. Genes reported to be mutated in these cancers were garnered from the literature (Table 2).

Example 3

Identification of Mutations in Tumor Tissues

[0053] We acquired tumors from 46 cancer patients in whom Pap smears were available. These included 24 patients with endometrial cancers and 22 with ovarian cancers; clinical and histopathologic features are listed in table S3.

[0054] Somatic mutations in the 46 tumors were identified through whole-exome sequencing as described above or through targeted sequencing of genes frequently mutated in the most common subtypes of ovarian or endometrial cancer (Table 2). Enrichment for these genes was achieved using a custom solid phase capture assay comprised of oligonucleotides ("capture probes") complementary to a panel of gene regions of interest. For the oncogenes, we only targeted their commonly mutated exons, whereas we targeted the entire coding regions of the tumor suppressor genes.

[0055] Illumina DNA sequencing libraries were generated from tumors and their matched non-neoplastic tissues, then captured with the assay described above. Following amplification by PCR, four to eight captured DNA libraries were sequenced per lane on an Illumina GA IIx instrument. In each of the 46 cases, we identified at least one somatic mutation (table S3) that was confirmed by an independent assay, as described below.

Example 4

Identification of Somatic Mutations in Pap Smears

[0056] In the liquid-based Pap smear technique in routine use today, the clinician inserts a small brush into the endocervical canal during a pelvic exam and rotates the brush so that it dislodges and adheres to loosely attached cells or cell fragments. The brush is then placed in a vial of fixative solution (e.g., ThinPrep). Some of the liquid from the vial is used to prepare a slide for cytological analysis or for purification of HPV DNA. In our study, an aliquot of the DNA purified from the liquid was used to assess for the presence of DNA from the cancers of the 46 patients described above. Preliminary studies showed that the fixed cells or cell fragments in the liquid, pelleted by centrifugation at 1,000 g for five minutes, contained >95% of the total DNA in the vial. We therefore purified DNA from the cell pellets when the amount of available liquid was greater than 3 mL (as occurs with some liquid-based Pap smear kits) and, for convenience, purified DNA from both the liquid and cells when smaller amounts of liquid were in the kit. In all cases, the purified DNA was of relatively high molecular weight (95%>5 kb). The average amount of DNA recovered from the 46 Pap smears was 49.3.+-.74.4 ng/ml (table S3).

[0057] We anticipated that, if present at all, the amount of DNA derived from neoplastic cells in the Pap smear fluid would be relatively small compared to the DNA derived from normal cells brushed from the endocervical canal. This necessitated the use of an analytic technique that could reliably identify a rare population of mutant alleles among a great excess of wild-type alleles. A modification of one of the Safe-SeqS (Safe-Sequencing System) procedures described in (30) was designed for this purpose (FIG. 2).

[0058] In brief, a limited number of PCR cycles was performed with a set of gene-specific primers. One of the primers contained 14 degenerate N bases (equal probability of being an A, C, G, or T) located 5' to its gene-specific sequence, and both primers contained sequences that permitted universal amplification in the next step. The 14 N's formed unique identifiers (UID) for each original template molecule. Subsequent PCR products generated with universal primers were purified and sequenced on an Illumina MiSeq instrument. If a mutation preexisted in a template molecule, that mutation should be present in every daughter molecule containing that UID, and such mutations are called "supermutants" (30). Mutations not occurring in the original templates, such as those occurring during the amplification steps or through errors in base calling, should not give rise to supermutants. The Safe-SeqS approach used here is capable of detecting 1 mutant template among 5,000 to 1,000,000 wild-type templates, depending on the amplicon and the position within the amplicon that is queried (30).

[0059] We designed Safe-SeqS primers (table S4) to detect at least one mutation from each of the 46 patients described in table S3. In the 24 Pap smears from patients with endometrial cancers, the mutation present in the tumor was identified in every case (100%). The median fraction of mutant alleles was 2.7%, and ranged from 0.01% to 78% (FIG. 3 and table S3). Amplifications of DNA from non-neoplastic tissues were used as negative controls in these experiments to define the detection limits of each queried mutation. In all cases, the fraction of mutant alleles was significantly different from the background mutation levels determined from the negative controls (P<0.001, binomial test). There was no obvious correlation between the fraction of mutant alleles and the histopathologic subtype or the stage of the cancer (FIG. 3 and table S3).

[0060] In two endometrial cancer cases, two mutations found in the tumor DNA were evaluated in the Pap smears (table S3). In both cases, the mutations were identified in DNA from the Pap smear (table S3). Moreover, the ratios between the mutant allele fractions of the two mutations in the Pap smears were correlated with those of the corresponding tumor samples. For example, in the Pap smear of case PAP 083 the mutant allele fractions for the CTNNB1 and PIK3CA mutations were 0.143% and 0.064%, respectively--a ratio of 2.2 (=0.14% to 0.064%). In the primary tumor from PAP 083, the corresponding ratio was 2.0 (79.5% to 39.5%).

[0061] Similar analysis of Pap smear DNA from ovarian cancer patients revealed detectable mutations in nine of the 22 patients (41%). The fraction of mutant alleles was smaller than in endometrial cancers (median of 0.49%, range 0.021% to 5.9%; see FIG. 3 and table S3). All but one of the cases with detectable mutations were epithelial tumors; the exception was a dysgerminoma, a malignant germ cell tumor of the ovary (table S3). As with endometrial cancers, there was no statistically significant correlation between the fraction of mutant alleles and histopathologic criteria. However, most ovarian cancers are detected only at an advanced stage, and this was reflected in the patients available in our cohort.

Example 5

A Genetic Test for Screening Purposes

[0062] The results described above document that mutant DNA molecules from most endometrial cancers and some ovarian cancers can be found in routinely collected Pap smears. However, in all 46 cases depicted in FIG. 3, a specific mutation was known to occur in the tumor, and an assay was subsequently designed to determine whether that mutation was also present in the corresponding Pap smears. In a screening setting, there obviously would be no known tumor prior to the test. We therefore designed a prototype test based on Safe-SeqS that could be used in a screening setting (FIG. 2).

[0063] This multiplexed approach included 50 primer pairs that amplified segments of 241 to 296 bp containing frequently mutated regions of DNA. The regions to be amplified were chosen from the results described in Section I and included exons from APC, AKT1, BRAF, CTNNB1, EGFR, FBXW7, KRAS, PIK3CA, PPP2R1A, PTEN, and TP53. In control experiments, 46 of the 50 amplicons were shown to provide information on a minimum of 2,500 templates; the number of templates sequenced can be determined directly from SafeSeqS-based sequencing (FIG. 2). Given the accuracy of SafeSeqS, this number was adequate to comfortably detect mutations existing in >0.1% of template molecules (30). The regions covered by these 46 amplicons (table S5), encompassing 10,257 bp, were predicted to be able to detect at least one mutation in >90% of either endometrial or ovarian cancers.

[0064] This test was applied to Pap smears of 14 cases--twelve endometrial and two ovarian--as well as 14 Pap smears collected from normal women. The 14 cancer cases were arbitrarily chosen from those which had mutant allele fractions >0.1% (table S3) and therefore above the detection limit of the multiplexed assay. In all 14 Pap smears from women with cancer, the mutation expected to be present (table S3) was identified (FIG. 4 and table S6). The fraction of mutant alleles in the multiplexed test was similar to that observed in the original analysis of the same samples using only one Safe-SeqS primer pair per amplicon (table S3 and table S6). Importantly, no mutations were detected in the 14 Pap smears from women without cancer (FIG. 4; see Materials and Methods).

Example 6

Materials and Methods

Patient Samples

[0065] All samples for this study were obtained using protocols approved by the Institutional Review Boards of The Johns Hopkins Medical Institutions (Baltimore, Md.), Memorial Sloan Kettering Cancer Center (New York, N.Y.), University of Sao Paulo (Sao Paulo, Brazil), and ILSbio, LLC (Chestertown, Md.). Demographic, clinical and pathologic staging data was collected for each case. All histopathology was centrally re-reviewed by board-certified pathologists. Staging was based on 2009 FIGO criteria (38).

[0066] Fresh-frozen tissue specimens of surgically resected neoplasms of the ovary and endometrium were analyzed by frozen section to assess neoplastic cellularity by a board-certified pathologist. Serial frozen sections were used to guide the trimming of Optimal Cutting Temperature (OCT) compound embedded frozen tissue blocks to enrich the fraction of neoplastic cells for DNA extraction.

[0067] Formalin-fixed paraffin embedded (FFPE) tissue samples were assessed by a board-certified pathologist (Propath LLC, Dallas, Tex.) for tumor cellularity and to demarcate area of high tumor cellularity. Tumor tissue from serial 10 micron sections on slides from the original tumor block were macrodissected with a razorblade to enrich the fraction of neoplastic cells for DNA extraction.

[0068] The source of normal DNA was matched whole blood or non-neoplastic normal adjacent tissue.

[0069] Liquid-based Pap smears were collected using cervical brushes and transport medium from Digene HC2 DNA Collection Device (Qiagen) or ThinPrep 2000 System (Hologic) and stored using the manufacturer's recommendations.

[0070] Unless otherwise indicated, all patient-related values are reported as mean.+-.1 standard deviation.

DNA Extraction

[0071] DNA was purified from tumor and normal tissue as well as liquid-based Pap Smears using an AllPrep kit (Qiagen) according to the manufacturer's instructions. DNA was purified from tumor tissue by adding 3 mL RLTM buffer (Qiagen) and then binding to an AllPrep DNA column (Qiagen) following the manufacturer's protocol. DNA was purified from Pap smear liquids by adding five volumes of RLTM buffer when the amount of liquid was less than 3 mL. When the amount of liquid was >3 mL, the cells and cell fragments were pelleted at 1,000.times.g for five minutes and the pellets were dissolved in 3 mL RLTM buffer. DNA was quantified in all cases with qPCR, employing the primers and conditions previously described (39).

Microsatellite Instability Testing

[0072] Microsatellite instability was detected using the MSI Analysis System (Promega), containing five mononucleotide repeats (BAT-25, BAT-26, NR-21, NR-24 and MONO-27) and two pentanucleotide repeat loci, per the manufacturer's instructions. Following amplification, the fluorescent PCR products were sized on an Applied Biosystems 3130 capillary electrophoresis instrument (Invitrogen). Tumor samples were designated as follows: MSI-high if two or more mononucleotides varied in length compared to the germline DNA; MSI-low if only one locus varied; and microsatellite stable (MSS) if there was no variation compared to the germline. Pentanucleotide loci confirmed identity in all cases.

Preparation of Illumina DNA Libraries and Capture for Exomic Sequencing

[0073] Preparation of Illumina genomic DNA libraries for exomic and targeted DNA captures was performed according to the manufacturer's recommendations. Briefly, 1-3 .mu.g of genomic DNA was used for library preparation using the TruSeqDNA Sample Preparation Kit (Illumina). The DNA was acoustically sheared (Covaris) to a target size of .about.200 bp. The fragments were subsequently end-repaired to convert overhangs into blunt ends. A single "A" nucleotide was then added to the 3' ends of blunt fragments to prevent them from later self-ligation; a corresponding "T" on the 3' end of adaptor molecules provided the complementary overhang. Following ligation to adaptors, the library was amplified with 8-14 cycles of PCR to ensure yields of 0.5 and 4 .mu.g for exomic and targeted gene captures, respectively.

[0074] Exomic capture was performed with the SureSelect Human Exome Kit V 4.0 (Agilent) according to the manufacturer's protocol, with the addition of TruSeq index-specific blocks in the hybridization mixture (AGATCGGAAGAGCACACGTCTGAACTCCAGTCAC-XXXXXX-ATCTCGTATGCCGTCTTCTGCTTGT (SEQ ID NO: 1), where the six base pair "XXXXXX" denotes one of 12 sample-specific indexes).

Targeted Gene Enrichment

[0075] Targeted gene enrichment was performed by modifications of previously described methods (40, 41). In brief, targeted regions of selected oncogenes and tumor suppressor genes were synthesized as oligonucleotide probes by Agilent Technologies. Probes of 36 bases were designed to capture both the plus and the minus strand of the DNA and had a 33-base overlap. The oligonucleotides were cleaved from the chip by incubating with 3 mL of 35% ammonium hydroxide at room temperate for five hours. The solution was transferred to two 2-ml tubes, dried under vacuum, and redissolved in 400 .mu.L of ribonuclease (RNase)- and deoxyribonuclease (DNase)-free water. Five microliters of the solution was used for PCR amplification with primers complementary to the 12-base sequence common to all probes: 5'-TGATCCCGCGACGA*C-3' (SEQ ID NO: 2) and 5'-GACCGCGACTCCAG*C-3' (SEQ ID NO: 3), with * indicating a phosphorothioate bond. The PCR products were purified with a MinElute Purification Column (Qiagen), end-repaired with End-IT DNA End-Repair Kit (Epicentre), and then purified with a MinElute Purification Column (Qiagen). The PCR products were ligated to form concatamers as described (40).

[0076] The major difference between the protocol described in (40, 41) and the one used in the present study involved the amplification of the ligated PCR products and the solid phase capture method. The modifications were as follows: 50 ng of ligated PCR product was amplified using the REPLI-g Midi Kit (Qiagen) with the addition of 2.5 nmol Biotin-dUTP (Roche) in a 27.5 .mu.L reaction. The reaction was incubated at 30.degree. C. for 16 hours, the polymerase was inactivated at 65.degree. C. for 3 mins. The amplified probes were purified with QiaQuick PCR Purification Columns (Qiagen). For capture, 4-5 .mu.g of library DNA was incubated with 1 .mu.g of the prepared probes in a hybridization mixture as previously described (40). The biotinylated probes and captured library sequences were subsequently purified using 500 .mu.g Dynabeads.RTM. MyOne Streptavidin (Invitrogen). After washing as per the manufacturer's recommendations, the captured sequences were eluted with 0.1 M NaOH and then neutralized with 1M Tris-HCl (pH 7.5). Neutralized DNA was desalted and concentrated using a QIAquick MinElute Column (Qiagen) in 20 .mu.L. The elute was amplified in a 100 .mu.L Phusion Hot Start II (Thermo Scientific) reaction containing 1.times. Phusion HF buffer, 0.25 mM dNTPs, 0.5 .mu.M each forward and reverse TruSeq primers, and 2 U polymerase with the following cycling conditions: 98.degree. C. for 30 s; 14 cycles of 98.degree. C. for 10 s, 60.degree. C. for 30 s, 72.degree. C. for 30 s; and 72.degree. C. for 5 min. The amplified pool containing enriched target sequences was purified using an Agencourt AMPure XP system (Beckman) and quantified using a 2100 Bioanalyzer (Agilent).

Next-Generation Sequencing and Somatic Mutation Identification

[0077] After capture of targeted sequences, paired-end sequencing using an Illumina GA IIx Genome Analyzer provided 2.times.75 base reads from each fragment. The sequence tags that passed filtering were aligned to the human genome reference sequence (hg18) and subsequent variant-calling analysis was performed using the ELANDv2 algorithm in the CASAVA 1.6 software (Illumina). Known polymorphisms recorded in dbSNP were removed from the analysis. Identification of high confidence mutations was performed as described previously (24).

Assessment of Low-Frequency Mutations

[0078] Primer Design. We attempted to design primer pairs to detect mutations in the 46 cancers described in the text. Primers were designed as described (30), using Primer3. (42) Sixty percent of the primers amplified the expected fragments; in the other 40%, a second or third set of primers had to be designed to reduce primer dimers or non-specific amplification.

[0079] Sequencing Library Preparation. Templates were amplified as described previously (30), with modifications that will be described in full elsewhere. In brief, each strand of each template molecule was encoded with a 14 base unique identifier (UID)--comprised of degenerate N bases (equal probability of being an A, C, G, or T)--using two to four cycles of amplicon-specific PCR (UID assignment PCR cycles, see FIG. 2). While both forward and reverse gene-specific primers contained universal tag sequences at their 5' ends--providing the primer binding sites for the second-round amplification--only the forward primer contained the UID, positioned between the 5' universal tag and the 3' gene-specific sequences (four N's were included in the reverse primer to facilitate sequencing done on paired-end libraries) (table S4). The UID assignment PCR cycles included Phusion Hot Start II (Thermo Scientific) in a 50 .mu.L reaction containing 1.times. Phusion HF buffer, 0.25 mM dNTPs, 0.5 .mu.M each of forward (containing 14 N's) and reverse primers, and 2 U of polymerase. Carryover of residual UID-containing primers to the second-round amplification, which can complicate template quantification (30), was minimized through exonuclease digestion at 370 C to degrade unincorporated primers and subsequent purification with AMPure XP beads (Beckman) and elution in 10 .mu.L TE (10 mM Tris-HCl, 1 mM EDTA, pH 8.0).

[0080] The eluted templates were amplified in a second-round PCR using primers containing the grafting sequences necessary for hybridization to the Illumina GA IIx flow cell at their 5' ends (FIG. 2) and two terminal 3' phosphorothioates to protect them from residual exonuclease activity (30). The reverse amplification primer additionally contained an index sequence between the 5 `grafting and 3` universal tag sequences to enable the PCR products from multiple individuals to be simultaneously analyzed in the same flow cell compartment of the sequencer (30). The second-round amplification reactions contained 1.times. Phusion HF buffer, 0.25 mM dNTPs, 0.5 .mu.M each of forward and reverse primers, and 2 U of polymerase in a total of 50 .mu.L. After an initial heat activation step at 980 C for 2 minutes, twenty-three cycles of PCR were performed using the following cycling conditions: 980 C for 10 s, 650 C for 15 s, and 720 C for 15 s. The multiplexed assay was performed in similar fashion utilizing six independent amplifications per sample with the primers described in table S5. The PCR products were purified using AMPure XP beads and used directly for sequencing on either the Illumina MiSeq or GA IIx instruments, with equivalent results.

[0081] Data Analysis. High quality sequence reads were analyzed as previously described. (30) Briefly, reads in which each of the 14 bases comprising the UID (representing one original template strand; see FIG. 2) had a quality score .gtoreq.15 were grouped by their UID. Only the UIDs supported by more than one read were retained for further analysis. The template-specific portion of the reads that contained the sequence of an expected amplification primer was matched to a reference sequence set using a custom script (available from the authors upon request). Artifactual mutations--introduced during the sample preparation and/or sequencing steps--were eliminated by requiring that >50% of reads sharing the same UID contained the identical mutation (a "supermutant;" see FIG. 2). For the 46 assays querying a single amplicon, we required that the fraction of mutant alleles was significantly different from the background mutation levels determined from a negative control (P<0.001, binomial test). As mutations are not known a priori in a screening environment, we used a more agnostic metric to detect mutations in the multiplexed assay. A threshold supermutant frequency was defined for each sample as equaling the mean frequency of all supermutants plus six standard deviations of the mean. Only supermutants exceeding this threshold were designated as mutations and reported in FIG. 4 and table S6.

REFERENCES

[0082] The disclosure of each reference cited is expressly incorporated herein. [0083] 1. M. Arbyn, A. Anttila, J. Jordan, G. Ronco, U. Schenck, N. Segnan, H. Wiener, A. Herbert, L. von Karsa, European Guidelines for Quality Assurance in Cervical Cancer Screening. Second edition--summary document. Ann Oncol 21, 448-458 (2010). [0084] 2. R. M. DeMay, Practical principles of cytopathology. (ASCP Press, Chicago, 2010), pp. xi, 402 p. [0085] 3. F. Bray, J. S. Ren, E. Masuyer, J. Ferlay, Global estimates of cancer prevalence for 27 sites in the adult population in 2008. Int J Cancer, (2012). [0086] 4. J. Ferlay, H. R. Shin, F. Bray, D. Forman, C. Mathers, D. M. Parkin, GLOBOCAN 2008 v2.0, Cancer Incidence and Mortality Worldwide (IARC CancerBase No. 10, Lyon, France, 2010). [0087] 5. S. B. Sams, H. S. Currens, S. S. Raab, Liquid-based Papanicolaou tests in endometrial carcinoma diagnosis. Performance, error root cause analysis, and quality improvement. Am J Clin Pathol 137, 248-254 (2012). [0088] 6. P. Smith, O. Bakos, G. Heimer, U. Ulmsten, Transvaginal ultrasound for identifying endometrial abnormality. Acta Obstet Gynecol Scand 70, 591-594 (1991). [0089] 7. H. Mitchell, G. Giles, G. Medley, Accuracy and survival benefit of cytological prediction of endometrial carcinoma on routine cervical smears. Int J Gynecol Pathol 12, 34-40 (1993). [0090] 8. K. J. Carlson, S. J. Skates, D. E. Singer, Screening for ovarian cancer. Ann Intern Med 121, 124-132 (1994). [0091] 9. H. Meden, A. Fattahi-Meibodi, CA 125 in benign gynecological conditions. Int J Biol Markers 13, 231-237 (1998). [0092] 10. S. S. Buys, E. Partridge, A. Black, C. C. Johnson, L. Lamerato, C. Isaacs, D. J. Reding, R. T. Greenlee, L. A. Yokochi, B. Kessel, E. D. Crawford, T. R. Church, G. L. Andriole, J. L. Weissfeld, M. N. Fouad, D. Chia, B. O'Brien, L. R. Ragard, J. D. Clapp, J. M. Rathmell, T. L. Riley, P. Hartge, P. F. Pinsky, C. S. Zhu, G. Izmirlian, B. S. Kramer, A. B. Miller, J. L. Xu, P. C. Prorok, J. K. Gohagan, C. D. Berg, Effect of screening on ovarian cancer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Randomized Controlled Trial. JAMA 305, 2295-2303 (2011). [0093] 11. ACOG Practice Bulletin. Clinical Management Guidelines for Obstetrician-Gynecologists. Number 60, March 2005. Pregestational diabetes mellitus. Obstet Gynecol 105, 675-685 (2005). [0094] 12. E. Partridge, A. R. Kreimer, R. T. Greenlee, C. Williams, J. L. Xu, T. R. Church, B. Kessel, C. C. Johnson, J. L. Weissfeld, C. Isaacs, G. L. Andriole, S. Ogden, L. R. Ragard, S. S. Buys, Results from four rounds of ovarian cancer screening in a randomized trial. Obstet Gynecol 113, 775-782 (2009). [0095] 13. American Cancer Society. Detailed guide: ovarian cancer--can ovarian cancer be found early? (Available at http://www.cancer.org/Cancer/OvarianCancer/DetailedGuide/ovarian-cancer-d- etection). [0096] 14. Screening for ovarian cancer: recommendation statement. U.S. Preventive Services Task Force. Am Fam Physician 71, 759-762 (2005). [0097] 15. ACOG Committee Opinion: number 280, December 2002. The role of the generalist obstetrician-gynecologist in the early detection of ovarian cancer. Obstet Gynecol 100, 1413-1416 (2002). [0098] 16. National Comprehensive Cancer Network Practice Guidelines in Oncology: ovarian cancer and genetic screening. (Available at http://www.nccn.org/professionals/physician_gls/PDF/genetics_screening.pd- f). [0099] 17. N. M. Lindor, G. M. Petersen, D. W. Hadley, A. Y. Kinney, S. Miesfeldt, K. H. Lu, P. Lynch, W. Burke, N. Press, Recommendations for the care of individuals with an inherited predisposition to Lynch syndrome: a systematic review. JAMA 296, 1507-1517 (2006). [0100] 18. J. P. Marques, L. B. Costa, A. P. Pinto, A. F. Lima, M. E. Duarte, A. P. Barbosa, P. L. Medeiros, Atypical glandular cells and cervical cancer: systematic review. Rev Assoc Med Bras 57, 234-238 (2011). [0101] 19. R. P. Insinga, A. G. Glass, B. B. Rush, Diagnoses and outcomes in cervical cancer screening: a population-based study. Am J Obstet Gynecol 191, 105-113 (2004). [0102] 20. K. E. Sharpless, P. F. Schnatz, S. Mandavilli, J. F. Greene, J. I. Sorosky, Dysplasia associated with atypical glandular cells on cervical cytology. Obstet Gynecol 105, 494-500 (2005). [0103] 21. C. P. DeSimone, M. E. Day, M. M. Tovar, C. S. Dietrich, 3rd, M. L. Eastham, S. C. Modesitt, Rate of pathology from atypical glandular cell Pap tests classified by the Bethesda 2001 nomenclature. Obstet Gynecol 107, 1285-1291 (2006). [0104] 22. C. S. Geier, M. Wilson, W. Creasman, Clinical evaluation of atypical glandular cells of undetermined significance. Am J Obstet Gynecol 184, 64-69 (2001). [0105] 23. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609-615 (2011). [0106] 24. S. Jones, T. L. Wang, M. Shih Ie, T. L. Mao, K. Nakayama, R. Roden, R. Glas, D. Slamon, L. A. Diaz, Jr., B. Vogelstein, K. W. Kinzler, V. E. Velculescu, N. Papadopoulos, Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228-231 (2010). [0107] 25. S. Jones, T. L. Wang, R. J. Kurman, K. Nakayama, V. E. Velculescu, B. Vogelstein, K. W. Kinzler, N. Papadopoulos, M. Shih Ie, Low-grade serous carcinomas of the ovary contain very few point mutations. J Pathol 226, 413-420 (2012). [0108] 26. S. A. Forbes, N. Bindal, S. Bamford, C. Cole, C. Y. Kok, D. Beare, M. Jia, R. Shepherd, K. Leung, A. Menzies, J. W. Teague, P. J. Campbell, M. R. Stratton, P. A. Futreal, COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res 39, D945-950 (2011). [0109] 27. E. Barrow, L. Robinson, W. Alduaij, A. Shenton, T. Clancy, F. Lalloo, J. Hill, D. G. Evans, Cumulative lifetime incidence of extracolonic cancers in Lynch syndrome: a report of 121 families with proven mutations. Clin Genet 75, 141-149 (2009). [0110] 28. K. Oda, D. Stokoe, Y. Taketani, F. McCormick, High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res 65, 10669-10673 (2005). [0111] 29. E. Kuhn, R. C. Wu, B. Guan, G. Wu, J. Zhang, Y. Wang, L. Song, X. Yuan, L. Wei, R. B. Roden, K. T. Kuo, K. Nakayama, B. Clarke, P. Shaw, N. Olvera, R. J. Kurman, D. A. Levine, T. L. Wang, I. M. Shih, Identification of Molecular Pathway Aberrations in Uterine Serous Carcinoma by Genome-wide Analyses. J Natl Cancer Inst, (2012). [0112] 30. I. Kinde, J. Wu, N. Papadopoulos, K. W. Kinzler, B. Vogelstein, Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA 108, 9530-9535 (2011). [0113] 31. H. F. Traut, G. N. Papanicolaou, Cancer of the Uterus: The Vaginal Smear in Its Diagnosis. Cal West Med 59, 121-122 (1943). [0114] 32. B. Vogelstein, K. W. Kinzler, Cancer genes and the pathways they control. Nat Med 10, 789-799 (2004). [0115] 33. J. M. Cooper, M. L. Erickson, Endometrial sampling techniques in the diagnosis of abnormal uterine bleeding. Obstet Gynecol Clin North Am 27, 235-244 (2000). [0116] 34. C. C. Gunderson, A. N. Fader, K. A. Carson, R. E. Bristow, Oncologic and reproductive outcomes with progestin therapy in women with endometrial hyperplasia and grade 1 adenocarcinoma: a systematic review. Gynecol Oncol 125, 477-482 (2012). [0117] 35. R. E. Bristow, R. S. Tomacruz, D. K. Armstrong, E. L. Trimble, F. J. Montz, Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol 20, 1248-1259 (2002). [0118] 36. M. H. Mayrand, E. Duarte-Franco, I. Rodrigues, S. D. Walter, J. Hanley, A. Ferenczy, S. Ratnam, F. Coutlee, E. L. Franco, Human papillomavirus DNA versus Papanicolaou screening tests for cervical cancer. N Engl J Med 357, 1579-1588 (2007). [0119] 37. P. Naucler, W. Ryd, S. Tornberg, A. Strand, G. Wadell, K. Elfgren, T. Radberg, B. Strander, B. Johansson, O. Forslund, B. G. Hansson, E. Rylander, J. Dillner, Human papillomavirus and Papanicolaou tests to screen for cervical cancer. N Engl J Med 357, 1589-1597 (2007). [0120] 38. S. Pecorelli, Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet 105, 103-104 (2009). [0121] 39. C. Rago, D. L. Huso, F. Diehl, B. Karim, G. Liu, N. Papadopoulos, Y. Samuels, V. E. Velculescu, B. Vogelstein, K. W. Kinzler, L. A. Diaz, Jr., Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Res 67, 9364-9370 (2007). [0122] 40. J. Wu, H. Matthaei, A. Maitra, M. Dal Molin, L. D. Wood, J. R. Eshleman, M. Goggins, M. I. Canto, R. D. Schulick, B. H. Edil, C. L. Wolfgang, A. P. Klein, L. A. Diaz, Jr., P. J. Allen, C. M. Schmidt, K. W. Kinzler, N. Papadopoulos, R. H. Hruban, B. Vogelstein, Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med 3, 92ra66 (2011). [0123] 41. J. He, J. Wu, Y. Jiao, N. Wagner-Johnston, R. F. Ambinder, L. A. Diaz, Jr., K. W. Kinzler, B. Vogelstein, N. Papadopoulos, IgH gene rearrangements as plasma biomarkers in Non-Hodgkin's lymphoma patients. Oncotarget 2, 178-185 (2011). [0124] 42. S. Rozen, H. Skaletsky, Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol 132, 365-386 (2000). [0125] 43. N. Howlader, A. M. Noone, M. Krapcho, N. Neyman, R. Aminou, S. F. Altekruse, C. L. Kosary, J. Ruhl, Z. Tatalovich, H. Cho, A. Mariotto, M. P. Eisner, D. R. Lewis, H. S. Chen, E. J. Feuer, K. A. Cronin, SEER Cancer Statistics Review, 1975-2009 (National Cancer Institute. Bethesda, Md., 2012). [0126] 44. A. Malpica, M. T. Deavers, K. Lu, D. C. Bodurka, E. N. Atkinson, D. M. Gershenson, E. G. Silva, Grading ovarian serous carcinoma using a two-tier system. Am J Surg Pathol 28, 496-504 (2004). [0127] 45, L. A. G. Ries, J. L. Young, G. E. Keel, M. P. Eisner, Y. D. Lin, M-J. Homer, SEER Survival Monograph: Cancer Survival Among Adults: US SEER Program, 1988-2001, Patient and Tumor Characteristics (NIH Pub. No. 07-6215. National Cancer Institute, Bethesda, Md., 2007). [0128] 46. C. A. Hamilton, M. K. Cheung, K. Osann, L. Chen, N. N. Teng, T. A. Longacre, M. A. Powell, M. R. Hendrickson, D. S. Kapp, J. K. Chan, Uterine papillary serous and clear cell carcinomas predict for poorer survival compared to grade 3 endometrioid corpus cancers. Br J Cancer 94, 642-646 (2006).

Sequence CWU 1

1

191165DNAArtificial Sequencecapture sequence 1agatcggaag agcacacgtc tgaactccag tcacnnnnnn atctcgtatg ccgtcttctg 60cttgt 65215DNAArtificial SequencePrimers 2tgatcccgcg acgac 15315DNAArtificial SequencePrimers 3gaccgcgact ccagc 15460DNAArtificial SequencePrimers 4cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngatcc aatccatttt tgttgtccag 60545DNAArtificial SequencePrimers 5cacacaggaa acagctatga ccatgtgagc aagaggcttt ggagt 45652DNAArtificial SequencePrimers 6cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggcca agacctgccc tg 52747DNAArtificial SequencePrimers 7cacacaggaa acagctatga ccatgtgctg tgactgcttg tagatgg 47856DNAArtificial SequencePrimers 8cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnccacc tcctcaaaca gctcaa 56946DNAArtificial SequencePrimers 9cacacaggaa acagctatga ccatgtgcag cttgcttagg tccact 461055DNAArtificial SequencePrimers 10cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggcc atctacaagc agtca 551149DNAArtificial SequencePrimers 11cacacaggaa acagctatga ccatgnnnnt caccatcgct atctgagca 491255DNAArtificial SequencePrimers 12cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncattg gtgatgattc gatgg 551345DNAArtificial SequencePrimers 13cacacaggaa acagctatga ccatgctgcc tggctcagaa ttcac 451454DNAArtificial SequencePrimers 14cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnccctt tcttgcggag attc 541545DNAArtificial SequencePrimers 15cacacaggaa acagctatga ccatgctact gggacggaac agctt 451655DNAArtificial SequencePrimers 16cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggaag agaatctccg caaga 551745DNAArtificial SequencePrimers 17cacacaggaa acagctatga ccatggcttc ttgtcctgct tgctt 451856DNAArtificial SequencePrimers 18cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggcct gtctcaatat cccaaa 561948DNAArtificial SequencePrimers 19cacacaggaa acagctatga ccatgttgtt tttctgtttc tccctctg 482054DNAArtificial SequencePrimers 20cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncaagg cactcttgcc tacg 542154DNAArtificial SequencePrimers 21cacacaggaa acagctatga ccatgcattt tcattatttt tattataagg cctg 542255DNAArtificial SequencePrimers 22cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnctgtg gtagtggcac cagaa 552349DNAArtificial SequencePrimers 23cacacaggaa acagctatga ccatgnnnna agcggctgtt agtcactgg 492454DNAArtificial SequencePrimers 24cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggccc ctgtcatctt ctgt 542545DNAArtificial SequencePrimers 25cacacaggaa acagctatga ccatggactt ggctgtccca gaatg 452658DNAArtificial SequencePrimers 26cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncaaga aatcgatagc atttgcag 582752DNAArtificial SequencePrimers 27cacacaggaa acagctatga ccatgtttat ttgctttgtc aagatcattt tt 522860DNAArtificial SequencePrimers 28cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnaggaa atatctgctt gctcattcaa 602954DNAArtificial SequencePrimers 29cacacaggaa acagctatga ccatggaagc agatactaag caggacacta tatc 543055DNAArtificial SequencePrimers 30cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttccc ttggattctg acaca 553145DNAArtificial SequencePrimers 31cacacaggaa acagctatga ccatgagcac cattcgttga taggc 453257DNAArtificial SequencePrimers 32cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncactg gcagcaacag tcttacc 573349DNAArtificial SequencePrimers 33cacacaggaa acagctatga ccatggattg cctttaccac tcagagaag 493457DNAArtificial SequencePrimers 34cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttgca gcaattcact gtaaagc 573555DNAArtificial SequencePrimers 35cacacaggaa acagctatga ccatgccgat gtaataaata tgcacatatc attac 553654DNAArtificial SequencePrimers 36cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncaagg cactcttgcc tacg 543754DNAArtificial SequencePrimers 37cacacaggaa acagctatga ccatgcattt tcattatttt tattataagg cctg 543857DNAArtificial SequencePrimers 38cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncactg gcagcaacag tcttacc 573949DNAArtificial SequencePrimers 39cacacaggaa acagctatga ccatggattg cctttaccac tcagagaag 494054DNAArtificial SequencePrimers 40cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncaagg cactcttgcc tacg 544154DNAArtificial SequencePrimers 41cacacaggaa acagctatga ccatgcattt tcattatttt tattataagg cctg 544258DNAArtificial SequencePrimers 42cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnagctc aaagcaattt ctacacga 584353DNAArtificial SequencePrimers 43cacacaggaa acagctatga ccatgnnnng cacttacctg tgactccata gaa 534460DNAArtificial SequencePrimers 44cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcttt tgatgacatt gcatacattc 604549DNAArtificial SequencePrimers 45cacacaggaa acagctatga ccatgnnnna ctccaaagcc tcttgctca 494655DNAArtificial SequencePrimers 46cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncagtt gcaaaccaga cctca 554749DNAArtificial SequencePrimers 47cacacaggaa acagctatga ccatgtgtgg agtatttgga tgacagaaa 494853DNAArtificial SequencePrimers 48cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngtggc aagtggctcc tga 534945DNAArtificial SequencePrimers 49cacacaggaa acagctatga ccatgnnnnc atgggcggca tgaac 455057DNAArtificial SequencePrimers 50cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnactgg cagcaacagt cttacct 575149DNAArtificial SequencePrimers 51cacacaggaa acagctatga ccatgnnnnc ctcaggattg cctttacca 495253DNAArtificial SequencePrimers 52cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnagtcc ggcttggagg atg 535345DNAArtificial SequencePrimers 53cacacaggaa acagctatga ccatgtcccc actcctcctt tcttc 455454DNAArtificial SequencePrimers 54cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggaaa gggacgaact ggtg 545549DNAArtificial SequencePrimers 55cacacaggaa acagctatga ccatgnnnnt agggcctctt gtgccttta 495655DNAArtificial SequencePrimers 56cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcttc tgtcccttcc cagaa 555749DNAArtificial SequencePrimers 57cacacaggaa acagctatga ccatgnnnng acttggctgt cccagaatg 495858DNAArtificial SequencePrimers 58cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcata ccaatttctc gattgagg 585949DNAArtificial SequencePrimers 59cacacaggaa acagctatga ccatgnnnnc ggctttttca acccttttt 496055DNAArtificial SequencePrimers 60cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggcc atctacaagc agtca 556149DNAArtificial SequencePrimers 61cacacaggaa acagctatga ccatgnnnnt caccatcgct atctgagca 496254DNAArtificial SequencePrimers 62cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngggac ggaacagctt tgag 546350DNAArtificial SequencePrimers 63cacacaggaa acagctatga ccatgnnnng cggagattct cttcctctgt 506454DNAArtificial SequencePrimers 64cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncggtg taggagctgc tggt 546548DNAArtificial SequencePrimers 65cacacaggaa acagctatga ccatgnnnna cccaggtcca gatgaagc 486655DNAArtificial SequencePrimers 66cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggcc atctacaagc agtca 556749DNAArtificial SequencePrimers 67cacacaggaa acagctatga ccatgnnnnt caccatcgct atctgagca 496853DNAArtificial SequencePrimers 68cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngtggc aagtggctcc tga 536945DNAArtificial SequencePrimers 69cacacaggaa acagctatga ccatgnnnnc atgggcggca tgaac 457057DNAArtificial SequencePrimers 70cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncgaaa agtgtttctg tcatcca 577149DNAArtificial SequencePrimers 71cacacaggaa acagctatga ccatgnnnng cccctcctca gcatcttat 497255DNAArtificial SequencePrimers 72cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncagtt gcaaaccaga cctca 557353DNAArtificial SequencePrimers 73cacacaggaa acagctatga ccatgnnnnt gtggagtatt tggatgacag aaa 537456DNAArtificial SequencePrimers 74cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttata tttccccatg ccaatg 567559DNAArtificial SequencePrimers 75cacacaggaa acagctatga ccatgnnnng gtgttttgaa atgtgtttta taatttaga 597655DNAArtificial SequencePrimers 76cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcttc tgtcccttcc cagaa 557749DNAArtificial SequencePrimers 77cacacaggaa acagctatga ccatgnnnng acttggctgt cccagaatg 497855DNAArtificial SequencePrimers 78cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaaaa agccgaaggt cacaa 557951DNAArtificial SequencePrimers 79cacacaggaa acagctatga ccatgnnnnc tcaagaagca gaaagggaag a 518060DNAArtificial SequencePrimers 80cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngatcc aatccatttt tgttgtccag 608149DNAArtificial SequencePrimers 81cacacaggaa acagctatga ccatgnnnnt gagcaagagg ctttggagt 498255DNAArtificial SequencePrimers 82cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgtga tgatggtgag gatgg 558357DNAArtificial SequencePrimers 83cacacaggaa acagctatga ccatgnnnnt ccactacaac tacatgtgta acagttc 578455DNAArtificial SequencePrimers 84cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnctccg tcatgtgctg tgact 558548DNAArtificial SequencePrimers 85cacacaggaa acagctatga ccatgnnnnc agctgtgggt tgattcca 488655DNAArtificial SequencePrimers 86cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggcc atctacaagc agtca 558749DNAArtificial SequencePrimers 87cacacaggaa acagctatga ccatgnnnnt caccatcgct atctgagca 498856DNAArtificial SequencePrimers 88cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnccaat ccatttttgt tgtcca 568949DNAArtificial SequencePrimers 89cacacaggaa acagctatga ccatgnnnnt gagcaagagg ctttggagt 499053DNAArtificial SequencePrimers 90cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnctgca cagggcaggt ctt 539153DNAArtificial SequencePrimers 91cacacaggaa acagctatga ccatgnnnnc tctgtctcct tcctcttcct aca 539255DNAArtificial SequencePrimers 92cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggcc atctacaagc agtca 559349DNAArtificial SequencePrimers 93cacacaggaa acagctatga ccatgnnnnt caccatcgct atctgagca 499455DNAArtificial SequencePrimers 94cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncacgc aaatttcctt ccact 559550DNAArtificial SequencePrimers 95cacacaggaa acagctatga ccatgnnnng gcctctgatt cctcactgat 509660DNAArtificial SequencePrimers 96cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngatcc aatccatttt tgttgtccag 609749DNAArtificial SequencePrimers 97cacacaggaa acagctatga ccatgnnnnt gagcaagagg ctttggagt 499857DNAArtificial SequencePrimers 98cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnactgg cagcaacagt cttacct 579949DNAArtificial SequencePrimers 99cacacaggaa acagctatga ccatgnnnnc ctcaggattg cctttacca 4910062DNAArtificial SequencePrimers 100gacgtaaaac gacggccagt nnnnnnnnnn nnnnaaagta acatttccaa tctactaatg 60ct 6210150DNAArtificial SequencePrimers 101cacacaggaa acagctatga ccatgnnnnt gagaaaatcc ctgttcccac 5010255DNAArtificial SequencePrimers 102cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggag cctcttacac ccagt 5510351DNAArtificial SequencePrimers 103cacacaggaa acagctatga ccatgnnnna aaaacactgg agtttcccaa a 5110455DNAArtificial SequencePrimers 104cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaaaa agccgaaggt cacaa 5510549DNAArtificial SequencePrimers 105cacacaggaa acagctatga ccatgnnnna tgcccccaag aatcctagt 4910655DNAArtificial SequencePrimers 106cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncatcc gtctactccc acgtt 5510748DNAArtificial SequencePrimers 107cacacaggaa acagctatga ccatgnnnna tcagctaccg ccaagtcc 4810855DNAArtificial SequencePrimers 108cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttcgt ccctttccag cttta 5510956DNAArtificial SequencePrimers 109cacacaggaa acagctatga ccatgnnnng gaatccagtg tttcttttaa atacct 5611054DNAArtificial SequencePrimers 110cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnaccag ccctgtcgtc tctc 5411151DNAArtificial SequencePrimers 111cacacaggaa acagctatga ccatgnnnng ccctgacttt caactctgtc t 5111259DNAArtificial SequencePrimers 112cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngcctc agattcactt ttatcacct 5911349DNAArtificial SequencePrimers 113cacacaggaa acagctatga ccatgnnnna ccaggagcca ttgtctttg 4911460DNAArtificial SequencePrimers 114cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcacc acattacata cttaccatgc 6011551DNAArtificial SequencePrimers 115cacacaggaa acagctatga ccatgnnnna aggggatctc ttcctgtatc c 5111655DNAArtificial SequencePrimers 116cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntctgg atcccagaag gtgag 5511749DNAArtificial SequencePrimers 117cacacaggaa acagctatga ccatgnnnng gccagtgctg tctctaagg 4911860DNAArtificial SequencePrimers 118cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngtcca caaaatgatt ctgaattagc 6011951DNAArtificial SequencePrimers 119cacacaggaa acagctatga ccatgnnnna cgatacacgt ctgcagtcaa c 5112062DNAArtificial SequencePrimers 120cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnacaga aatattttag aaagggacaa 60ca 6212151DNAArtificial SequencePrimers 121cacacaggaa acagctatga ccatgnnnna gaaaataccc cctccatcaa c 5112260DNAArtificial SequencePrimers 122cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngatcc aatccatttt tgttgtccag 6012353DNAArtificial SequencePrimers 123cacacaggaa acagctatga ccatgnnnnt ccaaactgac caaactgttc tta 5312457DNAArtificial SequencePrimers 124cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaaac ccaaaatctg ttttcca 5712550DNAArtificial SequencePrimers 125cacacaggaa acagctatga ccatgnnnng accataaccc accacagcta 5012655DNAArtificial SequencePrimers 126cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnctcct cccagagacc ccagt 5512749DNAArtificial SequencePrimers 127cacacaggaa acagctatga ccatgnnnnc atgagcgctg ctcagatag 4912854DNAArtificial SequencePrimers 128cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncctag gaaggcaggg gagt 5412949DNAArtificial SequencePrimers 129cacacaggaa acagctatga ccatgnnnnt gcatgttgct tttgtaccg 4913054DNAArtificial SequencePrimers 130cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnaccac ccgcacgtct gtag 5413148DNAArtificial SequencePrimers 131cacacaggaa acagctatga ccatgnnnna gccagtgctt gttgcttg 4813254DNAArtificial SequencePrimers 132cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncacac tgacgtgcct ctcc 5413349DNAArtificial SequencePrimers 133cacacaggaa acagctatga ccatgnnnnt tatctcccct ccccgtatc 4913455DNAArtificial SequencePrimers 134cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngattg tcagtgcgct tttcc 5513549DNAArtificial SequencePrimers 135cacacaggaa acagctatga ccatgnnnng ctaaggatgg gggttgcta 4913661DNAArtificial SequencePrimers 136cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgact ttaccttatc aatgtctcga 60a 6113748DNAArtificial SequencePrimers 137cacacaggaa acagctatga ccatgnnnng ctcgccccct taatctct 4813855DNAArtificial SequencePrimers 138cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggtac ttccggaacc tgtgc 5513949DNAArtificial SequencePrimers 139cacacaggaa acagctatga ccatgnnnnc cgagtcctag ggagaggag 4914058DNAArtificial SequencePrimers 140cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttgtt aatggtggct ttttgttt 5814154DNAArtificial SequencePrimers 141cacacaggaa acagctatga ccatgnnnna aatgatctaa caatgctctt ggac 5414258DNAArtificial SequencePrimers 142cgacgtaaaa cgacggccag tnnnnnnnnn nnnnncatgg aaggatgaga atttcaag 5814349DNAArtificial SequencePrimers 143cacacaggaa acagctatga ccatgnnnnt ggctacgacc cagttacca 4914455DNAArtificial SequencePrimers 144cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnaaacc gtagctgccc tggta 5514550DNAArtificial SequencePrimers 145cacacaggaa acagctatga ccatgnnnnt gactgctctt ttcacccatc 5014656DNAArtificial SequencePrimers 146cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntcatc ttgggcctgt gttatc 5614753DNAArtificial SequencePrimers 147cacacaggaa acagctatga ccatgnnnng atgagaggtg gatgggtagt agt 5314856DNAArtificial SequencePrimers 148cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttcag ggcatgaact acttgg 5614949DNAArtificial SequencePrimers 149cacacaggaa acagctatga ccatgnnnna tcctcccctg catgtgtta 4915056DNAArtificial SequencePrimers 150cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntccct cattgcactg tactcc 5615149DNAArtificial SequencePrimers 151cacacaggaa acagctatga ccatgnnnng gtgcttagtg gccatttgt 4915257DNAArtificial SequencePrimers 152cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntggtc tctcatggca ctgtact 5715352DNAArtificial SequencePrimers 153cacacaggaa acagctatga ccatgnnnna ttagcaattt gagggacaaa cc 5215462DNAArtificial SequencePrimers 154cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgtat ctcactcgat aatctggatg 60ac 6215555DNAArtificial SequencePrimers 155cacacaggaa acagctatga ccatgnnnnt gtcacattat aaagattcag gcaat 5515659DNAArtificial SequencePrimers 156cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnagttt gacagttaaa ggcatttcc 5915754DNAArtificial SequencePrimers 157cacacaggaa acagctatga ccatgnnnnt gtccttattt tggatatttc tccc 5415855DNAArtificial SequencePrimers 158cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaaga cccaggtcca gatga 5515948DNAArtificial SequencePrimers 159cacacaggaa acagctatga ccatgnnnna gaaatgcagg gggatacg 4816055DNAArtificial SequencePrimers 160cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaggc ataactgcac ccttg 5516149DNAArtificial SequencePrimers 161cacacaggaa acagctatga ccatgnnnng ggagtagatg gagcctggt 4916255DNAArtificial SequencePrimers 162cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgctg gatttggttc taggg 5516350DNAArtificial SequencePrimers 163cacacaggaa acagctatga ccatgnnnnt gccacttgca aagtttcttc 5016455DNAArtificial SequencePrimers 164cgacgtaaaa cgacggccag tnnnnnnnnn

nnnnnggaag aacctggacc ctctg 5516550DNAArtificial SequencePrimers 165cacacaggaa acagctatga ccatgnnnnt tttgagagtc gttcgattgc 5016655DNAArtificial SequencePrimers 166cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgcaa cctgttttgt gatgg 5516751DNAArtificial SequencePrimers 167cacacaggaa acagctatga ccatgnnnna ggaaaatgac aatgggaatg a 5116859DNAArtificial SequencePrimers 168cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgatt catcaggaga gcatttaag 5916952DNAArtificial SequencePrimers 169cacacaggaa acagctatga ccatgnnnnt tgtttttctg tttctccctc tg 5217057DNAArtificial SequencePrimers 170cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngatgg tatccatgtg gtgagtg 5717150DNAArtificial SequencePrimers 171cacacaggaa acagctatga ccatgnnnnt ttgtgatgct aaggctccat 5017253DNAArtificial SequencePrimers 172cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnactgc cttccgggtc act 5317349DNAArtificial SequencePrimers 173cacacaggaa acagctatga ccatgnnnna gcccaaccct tgtccttac 4917454DNAArtificial SequencePrimers 174cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaggc tgtcagtggg gaac 5417550DNAArtificial SequencePrimers 175cacacaggaa acagctatga ccatgnnnna acatatttgc atggggtgtg 5017655DNAArtificial SequencePrimers 176cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnccact gcatggttca ctctg 5517749DNAArtificial SequencePrimers 177cacacaggaa acagctatga ccatgnnnna tcctgtgagc gaagttcca 4917857DNAArtificial SequencePrimers 178cgacgtaaaa cgacggccag tnnnnnnnnn nnnnntgcct ctttctcttg gttttca 5717951DNAArtificial SequencePrimers 179cacacaggaa acagctatga ccatgnnnng gacctaagca agctgcagta a 5118060DNAArtificial SequencePrimers 180cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnacacc caatgaagaa tgtaattgat 6018154DNAArtificial SequencePrimers 181cacacaggaa acagctatga ccatgnnnng gttgtgtgta gatgtgagtt ttcc 5418259DNAArtificial SequencePrimers 182cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnttctg ttacattgtg cagagttca 5918350DNAArtificial SequencePrimers 183cacacaggaa acagctatga ccatgnnnnt ggttttgagc agagagatgg 5018455DNAArtificial SequencePrimers 184cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnggaag aagtcccaac catga 5518553DNAArtificial SequencePrimers 185cacacaggaa acagctatga ccatgnnnnt cacttttcct ttctacccaa aag 5318656DNAArtificial SequencePrimers 186cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngaatc tgcattccca gagaca 5618749DNAArtificial SequencePrimers 187cacacaggaa acagctatga ccatgnnnnc ctgtttccca tcctcttcc 4918856DNAArtificial SequencePrimers 188cgacgtaaaa cgacggccag tnnnnnnnnn nnnnngacac aaaacaggct caggac 5618949DNAArtificial SequencePrimers 189cacacaggaa acagctatga ccatgnnnna gaaaccaaag ccaaaagca 4919055DNAArtificial SequencePrimers 190cgacgtaaaa cgacggccag tnnnnnnnnn nnnnnccatg ggactgactt tctgc 5519149DNAArtificial SequencePrimers 191cacacaggaa acagctatga ccatgnnnnt catctggacc tgggtcttc 49

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed